1
|
Allouche-Arnon H, Montrazi ET, Subramani B, Fisler M, Spigel I, Frydman L, Mehlman T, Brandis A, Harris T, Bar-Shir A. A Genetically Engineered Reporter System Designed for 2H-MRI Allows Quantitative In Vivo Mapping of Transgene Expression. J Am Chem Soc 2024; 146:31624-31632. [PMID: 39527270 PMCID: PMC11583250 DOI: 10.1021/jacs.4c09572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
The ability to obtain quantitative spatial information on subcellular processes of deep tissues in vivo has been a long-standing challenge for molecular magnetic resonance imaging (MRI) approaches. This challenge remains even more so for quantifying readouts of genetically engineered MRI reporters. Here, we set to overcome this challenge with a molecular system designed to obtain quantitative 2H-MRI maps of a gene reporter. To this end, we synthesized deuterated thymidine, d3-thy, with three magnetically equivalent deuterons at its methyl group (-CD3), showing a singlet peak with a characteristic 2H-NMR frequency (δ = 1.7 ppm). The upfield 3.0 ppm offset from the chemical shift of the HDO signal (δ = 4.7 ppm) allows for spectrally resolving the two 2H NMR signals and quantifying the concentration of d3-thy based on the known concentration of a tissue's HDO. Following systemic administration of d3-thy, its accumulation as d3-thy monophosphate in cells expressing the human thymidine kinase 1 (hTK1) transgene was mapped with 2H-MRI. The data obtained in vivo show the ability to use the d3-thy/hTK1 pair as a reporter probe/reporter gene system to quantitatively map transgene expression with MRI. Relying on a structurally unmodified reporter probe (d3-thy) to image the expression of unmutated human protein (hTK1) shows the potential of molecular imaging with 2H-MRI to monitor gene reporters and other relevant biological targets.
Collapse
Affiliation(s)
- Hyla Allouche-Arnon
- Department
of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Elton T. Montrazi
- Department
of Chemical and Biological Physics, Weizmann
Institute of Science, Rehovot 7610001, Israel
| | - Balamurugan Subramani
- Department
of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michal Fisler
- Department
of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Inbal Spigel
- Department
of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Lucio Frydman
- Department
of Chemical and Biological Physics, Weizmann
Institute of Science, Rehovot 7610001, Israel
| | - Tevie Mehlman
- Department
of Life Sciences Core Facilities, Weizmann
Institute of Science, Rehovot 7610001, Israel
| | - Alexander Brandis
- Department
of Life Sciences Core Facilities, Weizmann
Institute of Science, Rehovot 7610001, Israel
| | - Talia Harris
- Department
of Chemical Research Support, Weizmann Institute
of Science, Rehovot 7610001, Israel
| | - Amnon Bar-Shir
- Department
of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
2
|
Lam B, Velasquez M, Ogiyama T, Godines K, Szu FY, Velasquez-Mao AJ, AlGhuraibawi W, Wang J, Messersmith PB, Vandsburger MH. Imaging of adeno-associated viral capsids for purposes of gene editing using CEST NMR/MRI. Magn Reson Med 2024; 92:792-806. [PMID: 38651648 PMCID: PMC11142879 DOI: 10.1002/mrm.30058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/19/2024] [Accepted: 02/03/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE Gene therapy using adeno-associated virus (AAV) vector-mediated gene delivery has undergone substantial growth in recent years with promising results in both preclinical and clinical studies, as well as emerging regulatory approval. However, the inability to quantify the efficacy of gene therapy from cellular delivery of gene-editing technology to specific functional outcomes is an obstacle for efficient development of gene therapy treatments. Building on prior works that used the CEST reporter gene lysine rich protein, we hypothesized that AAV viral capsids may generate endogenous CEST contrast from an abundance of surface lysine residues. METHODS NMR experiments were performed on isolated solutions of AAV serotypes 1-9 on a Bruker 800-MHz vertical scanner. In vitro experiments were performed for testing of CEST-NMR contrast of AAV2 capsids under varying pH, density, biological transduction stage, and across multiple serotypes and mixed biological media. Reverse transcriptase-polymerase chain reaction was used to quantify virus concentration. Subsequent experiments at 7 T optimized CEST saturation schemes for AAV contrast detection and detected AAV2 particles encapsulated in a biocompatible hydrogel administered in the hind limb of mice. RESULTS CEST-NMR experiments revealed CEST contrast up to 52% for AAV2 viral capsids between 0.6 and 0.8 ppm. CEST contrast generated by AAV2 demonstrated high levels of CEST contrast across a variety of chemical environments, concentrations, and saturation schemes. AAV2 CEST contrast displayed significant positive correlations with capsid density (R2 > 0.99, p < 0.001), pH (R2 = 0.97, p = 0.01), and viral titer per cell count (R2 = 0.92, p < 0.001). Transition to a preclinical field strength yielded up to 11.8% CEST contrast following optimization of saturation parameters. In vivo detection revealed statistically significant molecular contrast between viral and empty hydrogels using both mean values (4.67 ± 0.75% AAV2 vs. 3.47 ± 0.87% empty hydrogel, p = 0.02) and quantile analysis. CONCLUSION AAV2 viral capsids exhibit strong capacity as an endogenous CEST contrast agent and can potentially be used for monitoring and evaluation of AAV vector-mediated gene therapy protocols.
Collapse
Affiliation(s)
- Bonnie Lam
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
| | - Mark Velasquez
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
| | - Tomoko Ogiyama
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
| | - Kevin Godines
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
| | - Fan-Yun Szu
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
| | - A J Velasquez-Mao
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
| | | | - Jingshen Wang
- Division of Biostatistics, UC Berkeley, Berkeley, California, USA
| | - Phillip B Messersmith
- Department of Bioengineering, UC Berkeley, Berkeley, California, USA
- Department of Materials Science and Engineering, UC Berkeley, Berkeley, California, USA
- Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California, USA
| | | |
Collapse
|
3
|
Yun J, Huang Y, Miller ADC, Chang BL, Baldini L, Dhanabalan KM, Li E, Li H, Mukherjee A. Destabilized reporters for background-subtracted, chemically-gated, and multiplexed deep-tissue imaging. Chem Sci 2024; 15:11108-11121. [PMID: 39027298 PMCID: PMC11253201 DOI: 10.1039/d4sc00377b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/23/2024] [Indexed: 07/20/2024] Open
Abstract
Tracking gene expression in deep tissues requires genetic reporters that can be unambiguously detected using tissue penetrant techniques. Magnetic resonance imaging (MRI) is uniquely suited for this purpose; however, there is a dearth of reporters that can be reliably linked to gene expression with minimal interference from background tissue signals. Here, we present a conceptually new method for generating background-subtracted, drug-gated, multiplex images of gene expression using MRI. Specifically, we engineered chemically erasable reporters consisting of a water channel, aquaporin-1, fused to destabilizing domains, which are stabilized by binding to cell-permeable small-molecule ligands. We showed that this approach allows for highly specific detection of gene expression through differential imaging. In addition, by engineering destabilized aquaporin-1 variants with orthogonal ligand requirements, it is possible to distinguish distinct subpopulations of cells in mixed cultures. Finally, we demonstrated this approach in a mouse tumor model through differential imaging of gene expression with minimal background.
Collapse
Affiliation(s)
- Jason Yun
- Department of Chemistry, University of California Santa Barbara CA 93106 USA
| | - Yimeng Huang
- Department of Chemistry, University of California Santa Barbara CA 93106 USA
| | - Austin D C Miller
- Biomolecular Science and Engineering Graduate Program, University of California Santa Barbara CA 93106 USA
| | - Brandon L Chang
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Barbara CA 93106 USA
| | - Logan Baldini
- Department of Chemical Engineering, University of California Santa Barbara CA 93106 USA
| | - Kaamini M Dhanabalan
- Department of Chemical Engineering, University of California Santa Barbara CA 93106 USA
| | - Eugene Li
- Department of Chemical Engineering, University of California Santa Barbara CA 93106 USA
| | - Honghao Li
- Department of Chemistry, University of California Santa Barbara CA 93106 USA
| | - Arnab Mukherjee
- Department of Chemistry, University of California Santa Barbara CA 93106 USA
- Biomolecular Science and Engineering Graduate Program, University of California Santa Barbara CA 93106 USA
- Department of Chemical Engineering, University of California Santa Barbara CA 93106 USA
| |
Collapse
|
4
|
Huang Y, Chen X, Zhu Z, Mukherjee A. A Dual-Gene Reporter-Amplifier Architecture for Enhancing the Sensitivity of Molecular MRI by Water Exchange. Chembiochem 2024; 25:e202400087. [PMID: 38439618 PMCID: PMC11604348 DOI: 10.1002/cbic.202400087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 03/06/2024]
Abstract
The development of genetic reporters for magnetic resonance imaging (MRI) is essential for investigating biological functions in vivo. However, current MRI reporters have low sensitivity, making it challenging to create significant contrast against the tissue background, especially when only a small fraction of cells express the reporter. To overcome this limitation, we developed an approach for amplifying the sensitivity of molecular MRI by combining a chemogenetic contrast mechanism with a biophysical approach to increase water diffusion through the co-expression of a dual-gene construct comprising an organic anion transporting polypeptide, Oatp1b3, and a water channel, Aqp1. We first show that the expression of Aqp1 amplifies MRI contrast in cultured cells engineered to express Oatp1b3. We demonstrate that the contrast amplification is caused by Aqp1-driven increase in water exchange, which provides the gadolinium ions internalized by Oatp1b3-expressing cells with access to a larger water pool compared with exchange-limited conditions. We further show that our methodology allows cells to be detected using approximately 10-fold lower concentrations of gadolinium than that in the Aqp1-free scenario. Finally, we show that our approach enables the imaging of mixed-cell cultures containing a low fraction of Oatp1b3-labeled cells that are undetectable on the basis of Oatp1b3 expression alone.
Collapse
Affiliation(s)
| | - Xinyue Chen
- Department of Molecular, Cellular, and Developmental Biology
| | - Ziyue Zhu
- Department of Molecular, Cellular, and Developmental Biology
| | - Arnab Mukherjee
- Department of Chemistry
- Department of Molecular, Cellular, and Developmental Biology
- Department of Chemical Engineering
| |
Collapse
|
5
|
Huang Y, Chen X, Zhu Z, Mukherjee A. A dual-gene reporter-amplifier architecture for enhancing the sensitivity of molecular MRI by water exchange. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.22.576672. [PMID: 38328134 PMCID: PMC10849537 DOI: 10.1101/2024.01.22.576672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The development of genetic reporters for magnetic resonance imaging (MRI) is essential for investigating biological functions in intact animals. However, current MRI reporters have low sensitivity, making it challenging to create significant contrast against the tissue background, especially when only a small percentage of cells express the reporter. To overcome this limitation, we developed an approach that amplifies signals by co-expressing an MRI reporter gene, Oatp1b3, with a water channel, aquaporin-1 (Aqp1). We first show that the expression of Aqp1 amplifies the paramagnetic relaxation effect of Oatp1b3 by facilitating transmembrane water exchange. This mechanism provides Oatp1b3-expressing cells with access to a larger water pool compared with typical exchange-limited conditions. We further demonstrated that our methodology allows dual-labeled cells to be detected using approximately 10-fold lower concentrations of contrast agent than that in the Aqp1-free scenario. Finally, we show that our approach enables the imaging of mixed-cell populations containing a low fraction of Oatp1b3-labeled cells that are otherwise undetectable based on Oatp1b3 expression alone.
Collapse
Affiliation(s)
| | - Xinyue Chen
- Department of Molecular, Cellular, and Developmental Biology
| | - Ziyue Zhu
- Department of Molecular, Cellular, and Developmental Biology
| | - Arnab Mukherjee
- Department of Chemistry
- Department of Molecular, Cellular, and Developmental Biology
| |
Collapse
|
6
|
Fillion AJ, Bricco AR, Lee HD, Korenchan D, Farrar CT, Gilad AA. Development of a synthetic biosensor for chemical exchange MRI utilizing in silico optimized peptides. NMR IN BIOMEDICINE 2023; 36:e5007. [PMID: 37469121 PMCID: PMC11075521 DOI: 10.1002/nbm.5007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/21/2023]
Abstract
Chemical exchange saturation transfer (CEST) MRI has been identified as a novel alternative to classical diagnostic imaging. Over the last several decades, many studies have been conducted to determine possible CEST agents, such as endogenously expressed compounds or proteins, that can be utilized to produce contrast with minimally invasive procedures and reduced or non-existent levels of toxicity. In recent years there has been an increased interest in the generation of genetically engineered CEST contrast agents, typically based on existing proteins with CEST contrast or modified to produce CEST contrast. We have developed an in silico method for the evolution of peptide sequences to optimize CEST contrast and showed that these peptides could be combined to create de novo biosensors for CEST MRI. A single protein, superCESTide, was designed to be 198 amino acids. SuperCESTide was expressed in E. coli and purified with size exclusion chromatography. The magnetic transfer ratio asymmetry generated by superCESTide was comparable to levels seen in previous CEST reporters, such as protamine sulfate (salmon protamine) and human protamine. These data show that novel peptides with sequences optimized in silico for CEST contrast that utilize a more comprehensive range of amino acids can still produce contrast when assembled into protein units expressed in complex living environments.
Collapse
Affiliation(s)
- Adam J. Fillion
- Department of Chemical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Alexander R. Bricco
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Harvey D. Lee
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - David Korenchan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - Christian T. Farrar
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - Assaf A. Gilad
- Department of Chemical Engineering, Michigan State University, East Lansing, Michigan, USA
- Department of Radiology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
7
|
Nagar D, Vladimirov N, Farrar CT, Perlman O. Dynamic and rapid deep synthesis of chemical exchange saturation transfer and semisolid magnetization transfer MRI signals. Sci Rep 2023; 13:18291. [PMID: 37880343 PMCID: PMC10600114 DOI: 10.1038/s41598-023-45548-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/20/2023] [Indexed: 10/27/2023] Open
Abstract
Model-driven analysis of biophysical phenomena is gaining increased attention and utility for medical imaging applications. In magnetic resonance imaging (MRI), the availability of well-established models for describing the relations between the nuclear magnetization, tissue properties, and the externally applied magnetic fields has enabled the prediction of image contrast and served as a powerful tool for designing the imaging protocols that are now routinely used in the clinic. Recently, various advanced imaging techniques have relied on these models for image reconstruction, quantitative tissue parameter extraction, and automatic optimization of acquisition protocols. In molecular MRI, however, the increased complexity of the imaging scenario, where the signals from various chemical compounds and multiple proton pools must be accounted for, results in exceedingly long model simulation times, severely hindering the progress of this approach and its dissemination for various clinical applications. Here, we show that a deep-learning-based system can capture the nonlinear relations embedded in the molecular MRI Bloch-McConnell model, enabling a rapid and accurate generation of biologically realistic synthetic data. The applicability of this simulated data for in-silico, in-vitro, and in-vivo imaging applications is then demonstrated for chemical exchange saturation transfer (CEST) and semisolid macromolecule magnetization transfer (MT) analysis and quantification. The proposed approach yielded 63-99% acceleration in data synthesis time while retaining excellent agreement with the ground truth (Pearson's r > 0.99, p < 0.0001, normalized root mean square error < 3%).
Collapse
Affiliation(s)
- Dinor Nagar
- School of Electrical Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Nikita Vladimirov
- Department of Biomedical Engineering, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Christian T Farrar
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Or Perlman
- Department of Biomedical Engineering, Tel Aviv University, 6997801, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
8
|
He XY, Zhou YR, Mu T, Liao YF, Jiang L, Qin Y, Cai JH. Magnetic resonance imaging focused on the ferritin heavy chain 1 reporter gene detects neuronal differentiation in stem cells. Neural Regen Res 2023; 18:1563-1569. [PMID: 36571363 PMCID: PMC10075097 DOI: 10.4103/1673-5374.358608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The neuronal differentiation of mesenchymal stem cells offers a new strategy for the treatment of neurological disorders. Thus, there is a need to identify a noninvasive and sensitive in vivo imaging approach for real-time monitoring of transplanted stem cells. Our previous study confirmed that magnetic resonance imaging, with a focus on the ferritin heavy chain 1 reporter gene, could track the proliferation and differentiation of bone marrow mesenchymal stem cells that had been transduced with lentivirus carrying the ferritin heavy chain 1 reporter gene. However, we could not determine whether or when bone marrow mesenchymal stem cells had undergone neuronal differentiation based on changes in the magnetic resonance imaging signal. To solve this problem, we identified a neuron-specific enolase that can be differentially expressed before and after neuronal differentiation in stem cells. In this study, we successfully constructed a lentivirus carrying the neuron-specific enolase promoter and expressing the ferritin heavy chain 1 reporter gene; we used this lentivirus to transduce bone marrow mesenchymal stem cells. Cellular and animal studies showed that the neuron-specific enolase promoter effectively drove the expression of ferritin heavy chain 1 after neuronal differentiation of bone marrow mesenchymal stem cells; this led to intracellular accumulation of iron and corresponding changes in the magnetic resonance imaging signal. In summary, we established an innovative magnetic resonance imaging approach focused on the induction of reporter gene expression by a neuron-specific promoter. This imaging method can be used to noninvasively and sensitively detect neuronal differentiation in stem cells, which may be useful in stem cell-based therapies.
Collapse
Affiliation(s)
- Xiao-Ya He
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yi-Rui Zhou
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Tong Mu
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing; Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yi-Fan Liao
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics; Department of Nuclear Medicine, The Second Hospital of the Army Medical University, Chongqing, China
| | - Li Jiang
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Qin
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jin-Hua Cai
- Department of Radiology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
9
|
Yun J, Baldini L, Huang Y, Li E, Li H, Chacko AN, Miller AD, Wan J, Mukherjee A. Engineering ligand stabilized aquaporin reporters for magnetic resonance imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543364. [PMID: 37333371 PMCID: PMC10274688 DOI: 10.1101/2023.06.02.543364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Imaging transgene expression in live tissues requires reporters that are detectable with deeply penetrant modalities, such as magnetic resonance imaging (MRI). Here, we show that LSAqp1, a water channel engineered from aquaporin-1, can be used to create background-free, drug-gated, and multiplex images of gene expression using MRI. LSAqp1 is a fusion protein composed of aquaporin-1 and a degradation tag that is sensitive to a cell-permeable ligand, which allows for dynamic small molecule modulation of MRI signals. LSAqp1 improves specificity for imaging gene expression by allowing reporter signals to be conditionally activated and distinguished from the tissue background by difference imaging. In addition, by engineering destabilized aquaporin-1 variants with different ligand requirements, it is possible to image distinct cell types simultaneously. Finally, we expressed LSAqp1 in a tumor model and showed successful in vivo imaging of gene expression without background activity. LSAqp1 provides a conceptually unique approach to accurately measure gene expression in living organisms by combining the physics of water diffusion and biotechnology tools to control protein stability.
Collapse
Affiliation(s)
- Jason Yun
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
| | - Logan Baldini
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Yimeng Huang
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
| | - Eugene Li
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Honghao Li
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
| | - Asish N. Chacko
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
| | - Austin D.C. Miller
- Biomolecular Science and Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Jinyang Wan
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
| | - Arnab Mukherjee
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
- Biomolecular Science and Engineering, University of California, Santa Barbara, CA 93106, USA
- Biological Engineering, University of California, Santa Barbara, CA 93106, USA
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
10
|
Gilad AA, Bar-Shir A, Bricco AR, Mohanta Z, McMahon MT. Protein and peptide engineering for chemical exchange saturation transfer imaging in the age of synthetic biology. NMR IN BIOMEDICINE 2023; 36:e4712. [PMID: 35150021 PMCID: PMC10642350 DOI: 10.1002/nbm.4712] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 05/23/2023]
Abstract
At the beginning of the millennium, the first chemical exchange saturation transfer (CEST) contrast agents were bio-organic molecules. However, later, metal-based CEST agents (paraCEST agents) took center stage. This did not last too long as paraCEST agents showed limited translational potential. By contrast, the CEST field gradually became dominated by metal-free CEST agents. One branch of research stemming from the original work by van Zijl and colleagues is the development of CEST agents based on polypeptides. Indeed, in the last 2 decades, tremendous progress has been achieved in this field. This includes the design of novel peptides as biosensors, genetically encoded recombinant as well as synthetic reporters. This was a result of extensive characterization and elucidation of the theoretical requirements for rational designing and engineering of such agents. Here, we provide an extensive overview of the evolution of more precise protein-based CEST agents, review the rationalization of enzyme-substrate pairs as CEST contrast enhancers, discuss the theoretical considerations to improve peptide selectivity, specificity and enhance CEST contrast. Moreover, we discuss the strong influence of synthetic biology on the development of the next generation of protein-based CEST contrast agents.
Collapse
Affiliation(s)
- Assaf A. Gilad
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan, USA
- Department of Radiology, Michigan State University, East Lansing, Michigan, USA
| | - Amnon Bar-Shir
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander R. Bricco
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Zinia Mohanta
- Division of MR Research, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Michael T. McMahon
- Division of MR Research, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Maryland, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Bricco A, Miralavy I, Bo S, Perlman O, Korenchan DE, Farrar CT, McMahon MT, Banzhaf W, Gilad AA. A Genetic Programming Approach to Engineering MRI Reporter Genes. ACS Synth Biol 2023; 12:1154-1163. [PMID: 36947694 PMCID: PMC10128068 DOI: 10.1021/acssynbio.2c00648] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Indexed: 03/24/2023]
Abstract
Here we develop a mechanism of protein optimization using a computational approach known as "genetic programming". We developed an algorithm called Protein Optimization Engineering Tool (POET). Starting from a small library of literature values, the use of this tool allowed us to develop proteins that produce four times more MRI contrast than what was previously state-of-the-art. Interestingly, many of the peptides produced using POET were dramatically different with respect to their sequence and chemical environment than existing CEST producing peptides, and challenge prior understandings of how those peptides function. While existing algorithms for protein engineering rely on divergent evolution, POET relies on convergent evolution and consequently allows discovery of peptides with completely different sequences that perform the same function with as good or even better efficiency. Thus, this novel approach can be expanded beyond developing imaging agents and can be used widely in protein engineering.
Collapse
Affiliation(s)
- Alexander
R. Bricco
- Department
of Biomedical Engineering, Michigan State
University, East Lansing, Michigan 48823, United States
| | - Iliya Miralavy
- Department
of Computer Science & Engineering, Michigan
State University, East Lansing, Michigan 48823, United States
| | - Shaowei Bo
- The
Russell H. Morgan Department of Radiology and Radiological Sciences,
Division of MR Research, Johns Hopkins University
School of Medicine, Baltimore, Maryland 21218, United States
| | - Or Perlman
- Department
of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol
School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical
School, Boston, Massachusetts 02138, United States
| | - David E. Korenchan
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical
School, Boston, Massachusetts 02138, United States
| | - Christian T. Farrar
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical
School, Boston, Massachusetts 02138, United States
| | - Michael T. McMahon
- The
Russell H. Morgan Department of Radiology and Radiological Sciences,
Division of MR Research, Johns Hopkins University
School of Medicine, Baltimore, Maryland 21218, United States
| | - Wolfgang Banzhaf
- Department
of Computer Science & Engineering, Michigan
State University, East Lansing, Michigan 48823, United States
| | - Assaf A. Gilad
- Department
of Chemical Engineering and Materials Science, Michigan State University, East Lansing, Michigan 48823, United States
- Department
of Radiology, Michigan State University, East Lansing, Michigan 48823, United States
| |
Collapse
|
12
|
Fillion AJ, Bricco AR, Lee HD, Korenchan D, Farrar CT, Gilad AA. Development of a Synthetic Biosensor for Chemical Exchange MRI Utilizing In Silico Optimized Peptides. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.08.531737. [PMID: 37016672 PMCID: PMC10071792 DOI: 10.1101/2023.03.08.531737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Chemical Exchange Saturation Transfer (CEST) magnetic resonance imaging (MRI) has been identified as a novel alternative to classical diagnostic imaging. Over the last several decades, many studies have been conducted to determine possible CEST agents, such as endogenously expressed compounds or proteins, that can be utilized to produce contrast with minimally invasive procedures and reduced or non-existent levels of toxicity. In recent years there has been an increased interest in the generation of genetically engineered CEST contrast agents, typically based on existing proteins with CEST contrast or modified to produce CEST contrast. We have developed an in-silico method for the evolution of peptide sequences to optimize CEST contrast and showed that these peptides could be combined to create de novo biosensors for CEST MRI. A single protein, superCESTide 2.0, was designed to be 198 amino acids. SuperCESTide 2.0 was expressed in E. coli and purified with size-exclusion chromatography. The magnetic transfer ratio asymmetry (MTR asym ) generated by superCESTide 2.0 was comparable to levels seen in previous CEST reporters, such as protamine sulfate (salmon protamine, SP), Poly-L-Lysine (PLL), and human protamine (hPRM1). This data shows that novel peptides with sequences optimized in silico for CEST contrast that utilizes a more comprehensive range of amino acids can still produce contrast when assembled into protein units expressed in complex living environments.
Collapse
Affiliation(s)
- Adam J. Fillion
- Department of Chemical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Alexander R. Bricco
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Harvey D. Lee
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, USA
| | - David Korenchan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - Christian T. Farrar
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, MA, USA
| | - Assaf A. Gilad
- Department of Chemical Engineering, Michigan State University, East Lansing, Michigan, USA
- Department of Radiology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
13
|
Vladimirov N, Perlman O. Molecular MRI-Based Monitoring of Cancer Immunotherapy Treatment Response. Int J Mol Sci 2023; 24:3151. [PMID: 36834563 PMCID: PMC9959624 DOI: 10.3390/ijms24043151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Immunotherapy constitutes a paradigm shift in cancer treatment. Its FDA approval for several indications has yielded improved prognosis for cases where traditional therapy has shown limited efficiency. However, many patients still fail to benefit from this treatment modality, and the exact mechanisms responsible for tumor response are unknown. Noninvasive treatment monitoring is crucial for longitudinal tumor characterization and the early detection of non-responders. While various medical imaging techniques can provide a morphological picture of the lesion and its surrounding tissue, a molecular-oriented imaging approach holds the key to unraveling biological effects that occur much earlier in the immunotherapy timeline. Magnetic resonance imaging (MRI) is a highly versatile imaging modality, where the image contrast can be tailored to emphasize a particular biophysical property of interest using advanced engineering of the imaging pipeline. In this review, recent advances in molecular-MRI based cancer immunotherapy monitoring are described. Next, the presentation of the underlying physics, computational, and biological features are complemented by a critical analysis of the results obtained in preclinical and clinical studies. Finally, emerging artificial intelligence (AI)-based strategies to further distill, quantify, and interpret the image-based molecular MRI information are discussed in terms of perspectives for the future.
Collapse
Affiliation(s)
- Nikita Vladimirov
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Or Perlman
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
14
|
Abstract
MRI is a widely available clinical tool for cancer diagnosis and treatment monitoring. MRI provides excellent soft tissue imaging, using a wide range of contrast mechanisms, and can non-invasively detect tissue metabolites. These approaches can be used to distinguish cancer from normal tissues, to stratify tumor aggressiveness, and to identify changes within both the tumor and its microenvironment in response to therapy. In this review, the role of MRI in immunotherapy monitoring will be discussed and how it could be utilized in the future to address some of the unique clinical questions that arise from immunotherapy. For example, MRI could play a role in identifying pseudoprogression, mixed response, T cell infiltration, cell tracking, and some of the characteristic immune-related adverse events associated with these agents. The factors to be considered when developing MRI imaging biomarkers for immunotherapy will be reviewed. Finally, the advantages and limitations of each approach will be discussed, as well as the challenges for future clinical translation into routine clinical care. Given the increasing use of immunotherapy in a wide range of cancers and the ability of MRI to detect the microstructural and functional changes associated with successful response to immunotherapy, the technique has great potential for more widespread and routine use in the future for these applications.
Collapse
Affiliation(s)
- Doreen Lau
- Centre for Immuno-Oncology, University of Oxford, Oxford, UK
| | - Pippa G Corrie
- Department of Oncology, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
15
|
Yun J, Baldini M, Chowdhury R, Mukherjee A. Designing Protein-Based Probes for Sensing Biological Analytes with Magnetic Resonance Imaging. ANALYSIS & SENSING 2022; 2:e202200019. [PMID: 37409177 PMCID: PMC10321474 DOI: 10.1002/anse.202200019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Genetically encoded sensors provide unique advantages for monitoring biological analytes with molecular and cellular-level specificity. While sensors derived from fluorescent proteins represent staple tools in biological imaging, these probes are limited to optically accessible preparations owing to physical curbs on light penetration. In contrast to optical methods, magnetic resonance imaging (MRI) may be used to noninvasively look inside intact organisms at any arbitrary depth and over large fields of view. These capabilities have spurred the development of innovative methods to connect MRI readouts with biological targets using protein-based probes that are in principle genetically encodable. Here, we highlight the state-of-the-art in MRI-based biomolecular sensors, focusing on their physical mechanisms, quantitative characteristics, and biological applications. We also describe how innovations in reporter gene technology are creating new opportunities to engineer MRI sensors that are sensitive to dilute biological targets.
Collapse
Affiliation(s)
- Jason Yun
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
| | - Michelle Baldini
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Rochishnu Chowdhury
- Mechanical Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Arnab Mukherjee
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
- Biomolecular Science and Engineering, University of California, Santa Barbara, CA 93106, USA
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
- Center for BioEngineering, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
16
|
Sun J, Huang J, Bao G, Zheng H, Wang C, Wei J, Fu Y, Qiu J, Liao Y, Cai J. MRI detection of the malignant transformation of stem cells through reporter gene expression driven by a tumor-specific promoter. Stem Cell Res Ther 2021; 12:284. [PMID: 33980305 PMCID: PMC8117323 DOI: 10.1186/s13287-021-02359-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 04/27/2021] [Indexed: 01/10/2023] Open
Abstract
Background Existing evidence has shown that mesenchymal stem cells (MSCs) can undergo malignant transformation, which is a serious limitation of MSC-based therapies. Therefore, it is necessary to monitor malignant transformation of MSCs via a noninvasive imaging method. Although reporter gene-based magnetic resonance imaging (MRI) has been successfully applied to longitudinally monitor MSCs, this technique cannot distinguish the cells before and after malignant transformation. Herein, we investigated the feasibility of using a tumor-specific promoter to drive reporter gene expression for MRI detection of the malignant transformation of MSCs. Methods The reporter gene ferritin heavy chain (FTH1) was modified by adding a promoter from the tumor-specific gene progression elevated gene-3 (PEG3) and transduced into MSCs to obtain MSCs-PEG3-FTH1. Cells were induced to undergo malignant transformation via indirect coculture with C6 glioma cells, and these transformed cells were named MTMSCs-PEG3-FTH1. Western blot analysis of FTH1 expression, Prussian blue staining and transmission electron microscopy (TEM) to detect intracellular iron, and MRI to detect signal changes were performed before and after malignant transformation. Then, the cells before and after malignant transformation were inoculated subcutaneously into nude mice, and MRI was performed to observe the signal changes in the xenografts. Results After induction of malignant transformation, MTMSCs demonstrated tumor-like features in morphology, proliferation, migration, and invasion. FTH1 expression was significantly increased in MTMSCs-PEG3-FTH1 compared with MSCs-PEG3-FTH1. Prussian blue staining and TEM showed a large amount of iron particles in MTMSCs-PEG3-FTH1 but a minimal amount in MSCs-PEG3-FTH1. MRI demonstrated that the T2 value was significantly decreased in MTMSCs-PEG3-FTH1 compared with MSCs-PEG3-FTH1. In vivo, mass formation was observed in the MTMSCs-PEG3-FTH1 group but not the MSCs-PEG3-FTH1 group. T2-weighted MRI showed a significant signal decrease, which was correlated with iron accumulation in the tissue mass. Conclusions We developed a novel MRI model based on FTH1 reporter gene expression driven by the tumor-specific PEG3 promoter. This approach could be applied to sensitively detect the occurrence of MSC malignant transformation.
Collapse
Affiliation(s)
- Jun Sun
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China.,Department of Radiology, Chongqing University Central Hospital, Chongqing, 400014, China
| | - Jie Huang
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China
| | - Guangcheng Bao
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China
| | - Helin Zheng
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Cui Wang
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jie Wei
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China
| | - Yuanqiao Fu
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China
| | - Jiawen Qiu
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China.,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China.,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China
| | - Yifan Liao
- Department of Nuclear Medicine, Xinqiao Hospital affiliated with Third Military Medical University, Chongqing, 400037, China
| | - Jinhua Cai
- Department of Radiology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China. .,Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, 400014, China. .,Key Laboratory of Pediatrics in Chongqing, Chongqing, 400014, China. .,Chongqing International Science and Technology Cooperation Center for Child Development and Disorders, Chongqing, 400014, China.
| |
Collapse
|
17
|
Ahn HH, Carrington C, Hu Y, Liu HW, Ng C, Nam H, Park A, Stace C, West W, Mao HQ, Pomper MG, Ullman CG, Minn I. Nanoparticle-mediated tumor cell expression of mIL-12 via systemic gene delivery treats syngeneic models of murine lung cancers. Sci Rep 2021; 11:9733. [PMID: 33958660 PMCID: PMC8102550 DOI: 10.1038/s41598-021-89124-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 04/08/2021] [Indexed: 01/15/2023] Open
Abstract
Treatment of cancers in the lung remains a critical challenge in the clinic for which gene therapy could offer valuable options. We describe an effective approach through systemic injection of engineered polymer/DNA nanoparticles that mediate tumor-specific expression of a therapeutic gene, under the control of the cancer-selective progression elevated gene 3 (PEG-3) promoter, to treat tumors in the lungs of diseased mice. A clinically tested, untargeted, polyethylenimine carrier was selected to aid rapid transition to clinical studies, and a CpG-free plasmid backbone and coding sequences were used to reduce inflammation. Intravenous administration of nanoparticles expressing murine single-chain interleukin 12, under the control of PEG-3 promoter, significantly improved the survival of mice in both an orthotopic and a metastatic model of lung cancer with no marked symptoms of systemic toxicity. These outcomes achieved using clinically relevant nanoparticle components raises the promise of translation to human therapy.
Collapse
Affiliation(s)
- Hye-Hyun Ahn
- Division of Nuclear Medicine and Molecular Imaging, Russel H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | | | - Yizong Hu
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins University, School of Medicine, Baltimore, MD, 21287, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Heng-Wen Liu
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Christy Ng
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Hwanhee Nam
- Division of Nuclear Medicine and Molecular Imaging, Russel H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Andrew Park
- Division of Nuclear Medicine and Molecular Imaging, Russel H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
- AstraZeneca (MedImmune), One Medimmune Way, Gaithersburg, MD, 20878, USA
| | - Catherine Stace
- Cancer Targeting Systems, 1188 Centre Street, Newton Centre, MA, 02459, USA
- Platform First Ltd, 1 Station Road, Sutton, Cambridge, CB6 2RL, UK
| | - Will West
- Cancer Targeting Systems, 1188 Centre Street, Newton Centre, MA, 02459, USA
| | - Hai-Quan Mao
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins University, School of Medicine, Baltimore, MD, 21287, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Martin G Pomper
- Division of Nuclear Medicine and Molecular Imaging, Russel H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Christopher G Ullman
- Cancer Targeting Systems, 1188 Centre Street, Newton Centre, MA, 02459, USA.
- Paratopix Ltd., Bishop's Stortford, CM23 5JD, UK.
| | - Il Minn
- Division of Nuclear Medicine and Molecular Imaging, Russel H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA.
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA.
| |
Collapse
|
18
|
Wu AM. Protein Engineering for Molecular Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00045-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
19
|
Helfer BM, Bulte JW. Cell Surveillance Using Magnetic Resonance Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00042-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
20
|
Perlman O, Ito H, Gilad AA, McMahon MT, Chiocca EA, Nakashima H, Farrar CT. Redesigned reporter gene for improved proton exchange-based molecular MRI contrast. Sci Rep 2020; 10:20664. [PMID: 33244130 PMCID: PMC7692519 DOI: 10.1038/s41598-020-77576-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023] Open
Abstract
Reporter gene imaging allows for non-invasive monitoring of molecular processes in living cells, providing insights on the mechanisms underlying pathology and therapy. A lysine-rich protein (LRP) chemical exchange saturation transfer (CEST) MRI reporter gene has previously been developed and used to image tumor cells, cardiac viral gene transfer, and oncolytic virotherapy. However, the highly repetitive nature of the LRP reporter gene sequence leads to DNA recombination events and the expression of a range of truncated LRP protein fragments, thereby greatly limiting the CEST sensitivity. Here we report the use of a redesigned LRP reporter (rdLRP), aimed to provide excellent stability and CEST sensitivity. The rdLRP contains no DNA repeats or GC rich regions and 30% less positively charged amino-acids. RT-PCR of cell lysates transfected with rdLRP demonstrated a stable reporter gene with a single distinct band corresponding to full-length DNA. A distinct increase in CEST-MRI contrast was obtained in cell lysates of rdLRP transfected cells and in in vivo LRP expressing mouse brain tumors ([Formula: see text], n = 10).
Collapse
Affiliation(s)
- Or Perlman
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Suite 2301, Charlestown, MA, 02129, USA
| | - Hirotaka Ito
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Assaf A Gilad
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- The Institute of Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Department of Radiology, Michigan State University, East Lansing, MI, USA
| | - Michael T McMahon
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
- Division of MR Research, The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - E Antonio Chiocca
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hiroshi Nakashima
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Christian T Farrar
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, 149 13th Street, Suite 2301, Charlestown, MA, 02129, USA.
| |
Collapse
|
21
|
Zemerov SD, Roose BW, Farenhem KL, Zhao Z, Stringer MA, Goldman AR, Speicher DW, Dmochowski IJ. 129Xe NMR-Protein Sensor Reveals Cellular Ribose Concentration. Anal Chem 2020; 92:12817-12824. [PMID: 32897053 PMCID: PMC7649717 DOI: 10.1021/acs.analchem.0c00967] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Dysregulation of cellular ribose uptake can be indicative of metabolic abnormalities or tumorigenesis. However, analytical methods are currently limited for quantifying ribose concentration in complex biological samples. Here, we utilize the highly specific recognition of ribose by ribose-binding protein (RBP) to develop a single-protein ribose sensor detectable via a sensitive NMR technique known as hyperpolarized 129Xe chemical exchange saturation transfer (hyper-CEST). We demonstrate that RBP, with a tunable ribose-binding site and further engineered to bind xenon, enables the quantitation of ribose over a wide concentration range (nM to mM). Ribose binding induces the RBP "closed" conformation, which slows Xe exchange to a rate detectable by hyper-CEST. Such detection is remarkably specific for ribose, with the minimal background signal from endogenous sugars of similar size and structure, for example, glucose or ribose-6-phosphate. Ribose concentration was measured for mammalian cell lysate and serum, which led to estimates of low-mM ribose in a HeLa cell line. This highlights the potential for using genetically encoded periplasmic binding proteins such as RBP to measure metabolites in different biological fluids, tissues, and physiologic states.
Collapse
Affiliation(s)
- Serge D. Zemerov
- Department of Chemistry, University of Pennsylvania,
Philadelphia, PA 19104, USA
| | - Benjamin W. Roose
- Department of Chemistry, University of Pennsylvania,
Philadelphia, PA 19104, USA
| | - Kelsey L. Farenhem
- Department of Chemistry, University of Pennsylvania,
Philadelphia, PA 19104, USA
| | - Zhuangyu Zhao
- Department of Chemistry, University of Pennsylvania,
Philadelphia, PA 19104, USA
| | - Madison A. Stringer
- Department of Chemistry, University of Pennsylvania,
Philadelphia, PA 19104, USA
| | - Aaron R. Goldman
- Proteomics and Metabolomics Facility, The Wistar Institute,
Philadelphia, PA 19104, USA
| | - David W. Speicher
- Proteomics and Metabolomics Facility, The Wistar Institute,
Philadelphia, PA 19104, USA
- Molecular and Cellular Oncogenesis Program, The Wistar
Institute, Philadelphia, PA 19104, USA
| | - Ivan J. Dmochowski
- Department of Chemistry, University of Pennsylvania,
Philadelphia, PA 19104, USA
| |
Collapse
|
22
|
|
23
|
Spelkov AA, Goncharova EA, Savin AM, Kolpashchikov DM. Bifunctional RNA-Targeting Deoxyribozyme Nanodevice as a Potential Theranostic Agent. Chemistry 2020; 26:3489-3493. [PMID: 31943434 DOI: 10.1002/chem.201905528] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/09/2020] [Indexed: 01/14/2023]
Abstract
Theranostic approaches rely on simultaneous diagnostic of a disease and its therapy. Here, we designed a DNA nanodevice, which can simultaneously report the presence of a specific RNA target through an increase in fluorescence and cleave it. High selectivity of RNA target recognition under near physiological conditions was achieved. The proposed approach can become a basis for the design of DNA nanomachines and robots for diagnostics and therapy of viral infections, cancer, and genetic disorders.
Collapse
Affiliation(s)
- Aleksandr A Spelkov
- Laboratory of Solution Chemistry of Advanced Materials, and Technologies, ITMO University, Lomonosova St. 9, 191002, St. Petersburg, Russian Federation
| | - Ekaterina A Goncharova
- Laboratory of Solution Chemistry of Advanced Materials, and Technologies, ITMO University, Lomonosova St. 9, 191002, St. Petersburg, Russian Federation
| | - Artemii M Savin
- Laboratory of Solution Chemistry of Advanced Materials, and Technologies, ITMO University, Lomonosova St. 9, 191002, St. Petersburg, Russian Federation
| | - Dmitry M Kolpashchikov
- Laboratory of Solution Chemistry of Advanced Materials, and Technologies, ITMO University, Lomonosova St. 9, 191002, St. Petersburg, Russian Federation.,Chemistry Department, University of Central Florida, Orlando, 32816, Florida, USA.,Burnett School of Biomedical Sciences, University of Central Florida, Orlando, 32816, Florida, USA
| |
Collapse
|
24
|
Schuerle S, Furubayashi M, Soleimany AP, Gwisai T, Huang W, Voigt C, Bhatia SN. Genetic Encoding of Targeted Magnetic Resonance Imaging Contrast Agents for Tumor Imaging. ACS Synth Biol 2020; 9:392-401. [PMID: 31922737 DOI: 10.1021/acssynbio.9b00416] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Tumor-selective contrast agents have the potential to aid in the diagnosis and treatment of cancer using noninvasive imaging modalities such as magnetic resonance imaging (MRI). Such contrast agents can consist of magnetic nanoparticles incorporating functionalities that respond to cues specific to tumor environments. Genetically engineering magnetotactic bacteria to display peptides has been investigated as a means to produce contrast agents that combine the robust image contrast effects of magnetosomes with the transgenic-targeting peptides displayed on their surface. This work reports the first use of magnetic nanoparticles that display genetically encoded pH low insertion peptide (pHLIP), a long peptide intended to enhance MRI contrast by targeting the extracellular acidity associated with the tumors. To demonstrate the modularity of this versatile platform to incorporate diverse targeting ligands by genetic engineering, we also incorporated the cyclic αv integrin-binding peptide iRGD into separate magnetosomes. Specifically, we investigate their potential for enhanced binding and tumor imaging both in vitro and in vivo. Our experiments indicate that these tailored magnetosomes retain their magnetic properties, making them well suited as T2 contrast agents, while exhibiting an increased binding compared to the binding in wild-type magnetosomes.
Collapse
Affiliation(s)
- Simone Schuerle
- Institute for Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, CH-8092 Zurich, Switzerland
| | - Maiko Furubayashi
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Bioproduction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Sapporo 062-8517, Japan
| | - Ava P. Soleimany
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Harvard Graduate Program in Biophysics, Harvard University, Boston, Massachusetts 02115, United States
- Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Tinotenda Gwisai
- Institute for Translational Medicine, Department of Health Sciences and Technology, ETH Zurich, CH-8092 Zurich, Switzerland
| | - Wei Huang
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Christopher Voigt
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Sangeeta N. Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Marble Center for Cancer Nanomedicine, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
25
|
Ozbakir HF, Anderson NT, Fan KC, Mukherjee A. Beyond the Green Fluorescent Protein: Biomolecular Reporters for Anaerobic and Deep-Tissue Imaging. Bioconjug Chem 2020; 31:293-302. [PMID: 31794658 PMCID: PMC7033020 DOI: 10.1021/acs.bioconjchem.9b00688] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fluorescence imaging represents cornerstone technology for studying biological function at the cellular and molecular levels. The technology's centerpiece is a prolific collection of genetic reporters based on the green fluorescent protein (GFP) and related analogs. More than two decades of protein engineering have endowed the GFP repertoire with an incredible assortment of fluorescent proteins, allowing scientists immense latitude in choosing reporters tailored to various cellular and environmental contexts. Nevertheless, GFP and derivative reporters have specific limitations that hinder their unrestricted use for molecular imaging. These challenges have inspired the development of new reporter proteins and imaging mechanisms. Here, we review how these developments are expanding the frontiers of reporter gene techniques to enable nondestructive studies of cell function in anaerobic environments and deep inside intact animals-two important biological contexts that are fundamentally incompatible with the use of GFP-based reporters.
Collapse
Affiliation(s)
- Harun F. Ozbakir
- Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United States
| | - Nolan T. Anderson
- Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United States
| | - Kang-Ching Fan
- Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United States
| | - Arnab Mukherjee
- Department of Chemical Engineering, University of California, Santa Barbara, California 93106, United States
- Department of Chemistry, University of California, Santa Barbara, California 93106, United States
- Neuroscience Research Institute, University of California, Santa Barbara, California 93106, United States
- Center for Bioengineering, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
26
|
Hu Y, He Z, Hao Y, Gong L, Pang M, Howard GP, Ahn HH, Brummet M, Chen K, Liu HW, Ke X, Zhu J, Anderson CF, Cui H, Ullman CG, Carrington CA, Pomper MG, Seo JH, Mittal R, Minn I, Mao HQ. Kinetic Control in Assembly of Plasmid DNA/Polycation Complex Nanoparticles. ACS NANO 2019; 13:10161-10178. [PMID: 31503450 DOI: 10.1021/acsnano.9b03334.s004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Polyelectrolyte complex (PEC) nanoparticles assembled from plasmid DNA (pDNA) and polycations such as linear polyethylenimine (lPEI) represent a major nonviral delivery vehicle for gene therapy tested thus far. Efforts to control the size, shape, and surface properties of pDNA/polycation nanoparticles have been primarily focused on fine-tuning the molecular structures of the polycationic carriers and on assembly conditions such as medium polarity, pH, and temperature. However, reproducible production of these nanoparticles hinges on the ability to control the assembly kinetics, given the nonequilibrium nature of the assembly process and nanoparticle composition. Here we adopt a kinetically controlled mixing process, termed flash nanocomplexation (FNC), that accelerates the mixing of pDNA solution with polycation lPEI solution to match the PEC assembly kinetics through turbulent mixing in a microchamber. This achieves explicit control of the kinetic conditions for pDNA/lPEI nanoparticle assembly, as demonstrated by the tunability of nanoparticle size, composition, and pDNA payload. Through a combined experimental and simulation approach, we prepared pDNA/lPEI nanoparticles having an average of 1.3 to 21.8 copies of pDNA per nanoparticle and average size of 35 to 130 nm in a more uniform and scalable manner than bulk mixing methods. Using these nanoparticles with defined compositions and sizes, we showed the correlation of pDNA payload and nanoparticle formulation composition with the transfection efficiencies and toxicity in vivo. These nanoparticles exhibited long-term stability at -20 °C for at least 9 months in a lyophilized formulation, validating scalable manufacture of an off-the-shelf nanoparticle product with well-defined characteristics as a gene medicine.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Christopher G Ullman
- Cancer Targeting Systems , Chesterford Research Park , Cambridge , CB10 1XL , U.K
| | | | | | | | | | | | | |
Collapse
|
27
|
Hu Y, He Z, Hao Y, Liu HW, Gong L, Howard G, Ahn HH, Brummet M, Ke X, Anderson C, Seo JH, Zhu J, Chen K, Pang Wan Rion M, Cui H, Ullman CG, Carrington CA, Pomper MG, Mittal R, Minn I, Mao HQ. Kinetic Control in Assembly of Plasmid DNA/Polycation Complex Nanoparticles. ACS NANO 2019; 13:10161-10178. [PMID: 31503450 PMCID: PMC7293580 DOI: 10.1021/acsnano.9b03334] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Polyelectrolyte complex (PEC) nanoparticles assembled from plasmid DNA (pDNA) and polycations such as linear polyethylenimine (lPEI) represent a major nonviral delivery vehicle for gene therapy tested thus far. Efforts to control the size, shape, and surface properties of pDNA/polycation nanoparticles have been primarily focused on fine-tuning the molecular structures of the polycationic carriers and on assembly conditions such as medium polarity, pH, and temperature. However, reproducible production of these nanoparticles hinges on the ability to control the assembly kinetics, given the nonequilibrium nature of the assembly process and nanoparticle composition. Here we adopt a kinetically controlled mixing process, termed flash nanocomplexation (FNC), that accelerates the mixing of pDNA solution with polycation lPEI solution to match the PEC assembly kinetics through turbulent mixing in a microchamber. This achieves explicit control of the kinetic conditions for pDNA/lPEI nanoparticle assembly, as demonstrated by the tunability of nanoparticle size, composition, and pDNA payload. Through a combined experimental and simulation approach, we prepared pDNA/lPEI nanoparticles having an average of 1.3 to 21.8 copies of pDNA per nanoparticle and average size of 35 to 130 nm in a more uniform and scalable manner than bulk mixing methods. Using these nanoparticles with defined compositions and sizes, we showed the correlation of pDNA payload and nanoparticle formulation composition with the transfection efficiencies and toxicity in vivo. These nanoparticles exhibited long-term stability at -20 °C for at least 9 months in a lyophilized formulation, validating scalable manufacture of an off-the-shelf nanoparticle product with well-defined characteristics as a gene medicine.
Collapse
Affiliation(s)
- Yizong Hu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Zhiyu He
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yue Hao
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Heng-wen Liu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Like Gong
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Gregory Howard
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hye-Hyun Ahn
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Mary Brummet
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Xiyu Ke
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Caleb Anderson
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jung-Hee Seo
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jinchang Zhu
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kuntao Chen
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Marion Pang Wan Rion
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Honggang Cui
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | | - Martin G. Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Rajat Mittal
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Hai-Quan Mao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Correspondence should be addressed to Dr. Hai-Quan Mao: 3400 N. Charles Street, Croft Hall 100, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA.
| |
Collapse
|
28
|
Rotz MW, Holbrook RJ, MacRenaris KW, Meade TJ. A Markedly Improved Synthetic Approach for the Preparation of Multifunctional Au-DNA Nanoparticle Conjugates Modified with Optical and MR Imaging Probes. Bioconjug Chem 2018; 29:3544-3549. [PMID: 30193061 DOI: 10.1021/acs.bioconjchem.8b00504] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We describe a new, and vastly superior approach for labeling spherical nucleic acid conjugates (SNAs) with diagnostic probes. SNAs have been shown to provide the unique ability to traverse the cell membrane and deliver surface conjugated DNA into cells while preserving the DNA from nuclease degradation. Our previous work on preparing diagnostically labeled SNAs was labor intensive, relatively low yielding, and costly. Here, we describe a straightforward and facile preparation for labeling SNAs with optical and MR imaging probes with significantly improved physical properties. The synthesis of Gd(III) labeled DNA Au nanoparticle conjugates is achieved by sequential conjugation of 3'-thiol-modified oligonucleotides and cofunctionalization of the particle surface with the subsequent addition of 1,2 diothiolate modified chelates of Gd(III) (abbreviated: DNA-GdIII@AuNP). This new generation of SNA conjugates has a 2-fold increase of DNA labeling and a 1.4-fold increase in Gd(III) loading compared to published constructs. Furthermore, the relaxivity ( r1) is observed to increase 4.5-fold compared to the molecular dithiolane-Gd(III) complex, and 1.4-fold increase relative to previous particle constructs where the Gd(III) complexes were conjugated to the oligonucleotides rather than directly to the Au particle. Importantly, this simplified approach (2 steps) exploits the advantages of previous Gd(III) labeled SNA platforms; however, this new approach is scalable and eliminates modification of DNA for attaching the contrast agent, and the particles exhibit improved cell labeling.
Collapse
Affiliation(s)
- Matthew W Rotz
- Department of Chemistry, Molecular Biosciences, Neurobiology, Biomedical Engineering, and Radiology , Northwestern University , 2145 Sheridan Road , Evanston , Illinois 60208-3113 , United States
| | - Robert J Holbrook
- Department of Chemistry, Molecular Biosciences, Neurobiology, Biomedical Engineering, and Radiology , Northwestern University , 2145 Sheridan Road , Evanston , Illinois 60208-3113 , United States
| | - Keith W MacRenaris
- Department of Chemistry, Molecular Biosciences, Neurobiology, Biomedical Engineering, and Radiology , Northwestern University , 2145 Sheridan Road , Evanston , Illinois 60208-3113 , United States
| | - Thomas J Meade
- Department of Chemistry, Molecular Biosciences, Neurobiology, Biomedical Engineering, and Radiology , Northwestern University , 2145 Sheridan Road , Evanston , Illinois 60208-3113 , United States
| |
Collapse
|
29
|
Non-invasive detection of adeno-associated viral gene transfer using a genetically encoded CEST-MRI reporter gene in the murine heart. Sci Rep 2018; 8:4638. [PMID: 29545551 PMCID: PMC5854573 DOI: 10.1038/s41598-018-22993-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 03/05/2018] [Indexed: 01/02/2023] Open
Abstract
Research into gene therapy for heart failure has gained renewed interest as a result of improved safety and availability of adeno-associated viral vectors (AAV). While magnetic resonance imaging (MRI) is standard for functional assessment of gene therapy outcomes, quantitation of gene transfer/expression relies upon tissue biopsy, fluorescence or nuclear imaging. Imaging of gene expression through the use of genetically encoded chemical exchange saturation transfer (CEST)-MRI reporter genes could be combined with clinical cardiac MRI methods to comprehensively probe therapeutic gene expression and subsequent outcomes. The CEST-MRI reporter gene Lysine Rich Protein (LRP) was cloned into an AAV9 vector and either administered systemically via tail vein injection or directly injected into the left ventricular free wall of mice. Longitudinal in vivo CEST-MRI performed at days 15 and 45 after direct injection or at 1, 60 and 90 days after systemic injection revealed robust CEST contrast in myocardium that was later confirmed to express LRP by immunostaining. Ventricular structure and function were not impacted by expression of LRP in either study arm. The ability to quantify and link therapeutic gene expression to functional outcomes can provide rich data for further development of gene therapy for heart failure.
Collapse
|
30
|
|
31
|
Slavcev RA, Sum CH, St Jean J, Huh H, Nafissi N. Specific Systems for Evaluation. EXPERIENTIA SUPPLEMENTUM (2012) 2018; 110:99-123. [PMID: 30536228 DOI: 10.1007/978-3-319-78259-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Fluorescent-based visualization techniques have long been used to monitor biological activity. This chapter explores the delivery of reporter genes as a means to assay and track activity in biological systems. Bioluminescence is the production of light due to biochemical processes. By encoding genes for bioluminescence, biological processes can be visualized based on gene expression. This chapter also discusses the primary applications of bioluminescence as seen through bioluminescent imaging techniques, flow cytometry, and PCR-based methods of gene detection. These techniques are described in terms of researching gene expression, cancer therapy, and protein interactions.
Collapse
Affiliation(s)
| | - Chi Hong Sum
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| | - Jesse St Jean
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| | - Haein Huh
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| | - Nafiseh Nafissi
- University of Waterloo, School of Pharmacy, Waterloo, ON, Canada
| |
Collapse
|
32
|
Mukherjee A, Davis HC, Ramesh P, Lu GJ, Shapiro MG. Biomolecular MRI reporters: Evolution of new mechanisms. PROGRESS IN NUCLEAR MAGNETIC RESONANCE SPECTROSCOPY 2017; 102-103:32-42. [PMID: 29157492 PMCID: PMC5726449 DOI: 10.1016/j.pnmrs.2017.05.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 05/23/2017] [Accepted: 05/28/2017] [Indexed: 05/08/2023]
Abstract
Magnetic resonance imaging (MRI) is a powerful technique for observing the function of specific cells and molecules inside living organisms. However, compared to optical microscopy, in which fluorescent protein reporters are available to visualize hundreds of cellular functions ranging from gene expression and chemical signaling to biomechanics, to date relatively few such reporters are available for MRI. Efforts to develop MRI-detectable biomolecules have mainly focused on proteins transporting paramagnetic metals for T1 and T2 relaxation enhancement or containing large numbers of exchangeable protons for chemical exchange saturation transfer. While these pioneering developments established several key uses of biomolecular MRI, such as imaging of gene expression and functional biosensing, they also revealed that low molecular sensitivity poses a major challenge for broader adoption in biology and medicine. Recently, new classes of biomolecular reporters have been developed based on alternative contrast mechanisms, including enhancement of spin diffusivity, interactions with hyperpolarized nuclei, and modulation of blood flow. These novel reporters promise to improve sensitivity and enable new forms of multiplexed and functional imaging.
Collapse
Affiliation(s)
- Arnab Mukherjee
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Hunter C Davis
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Pradeep Ramesh
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - George J Lu
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Mikhail G Shapiro
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
33
|
Bar-Shir A, Alon L, Korrer MJ, Lim HS, Yadav NN, Kato Y, Pathak AP, Bulte JWM, Gilad AA. Quantification and tracking of genetically engineered dendritic cells for studying immunotherapy. Magn Reson Med 2017; 79:1010-1019. [PMID: 28480589 DOI: 10.1002/mrm.26708] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 03/15/2017] [Accepted: 03/18/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE Genetically encoded reporters can assist in visualizing biological processes in live organisms and have been proposed for longitudinal and noninvasive tracking of therapeutic cells in deep tissue. Cells can be labeled in situ or ex vivo and followed in live subjects over time. Nevertheless, a major challenge for reporter systems is to identify the cell population that actually expresses an active reporter. METHODS We have used a nucleoside analog, pyrrolo-2'-deoxycytidine, as an imaging probe for the putative reporter gene, Drosophila melanogaster 2'-deoxynucleoside kinase. Bioengineered cells were imaged in vivo in animal models of brain tumor and immunotherapy using chemical exchange saturation transfer MRI. The number of transduced cells was quantified by flow cytometry based on the optical properties of the probe. RESULTS We performed a comparative analysis of six different cell lines and demonstrate utility in a mouse model of immunotherapy. The proposed technology can be used to quantify the number of labeled cells in a given region, and moreover is sensitive enough to detect less than 10,000 cells. CONCLUSION This unique technology that enables efficient selection of labeled cells followed by in vivo monitoring with both optical and MRI. Magn Reson Med 79:1010-1019, 2018. © 2017 International Society for Magnetic Resonance in Medicine.
Collapse
Affiliation(s)
- Amnon Bar-Shir
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lina Alon
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael J Korrer
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland, USA
| | - Hong Seo Lim
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nirbhay N Yadav
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | - Yoshinori Kato
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Arvind P Pathak
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeff W M Bulte
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA.,Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Assaf A Gilad
- The Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| |
Collapse
|
34
|
McMahon MT, Gilad AA. Cellular and Molecular Imaging Using Chemical Exchange Saturation Transfer. Top Magn Reson Imaging 2017; 25:197-204. [PMID: 27748713 DOI: 10.1097/rmr.0000000000000105] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Chemical exchange saturation transfer (CEST) is a powerful new tool well suited for molecular imaging. This technology enables the detection of low concentration probes through selective labeling of rapidly exchanging protons or other spins on the probes. In this review, we will highlight the unique features of CEST imaging technology and describe the different types of CEST agents that are suited for molecular imaging studies, including CEST theranostic agents, CEST reporter genes, and CEST environmental sensors.
Collapse
Affiliation(s)
- Michael T McMahon
- *F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute †The Russell H. Morgan Department of Radiology and Radiological Sciences, Division of MR Research ‡Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD
| | | |
Collapse
|
35
|
Cho IK, Wang S, Mao H, Chan AWS. Genetic engineered molecular imaging probes for applications in cell therapy: emphasis on MRI approach. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2016; 6:234-261. [PMID: 27766183 PMCID: PMC5069277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 08/31/2016] [Indexed: 06/06/2023]
Abstract
Recent advances in stem cell-based regenerative medicine, cell replacement therapy, and genome editing technologies (i.e. CRISPR-Cas 9) have sparked great interest in in vivo cell monitoring. Molecular imaging promises a unique approach to noninvasively monitor cellular and molecular phenomena, including cell survival, migration, proliferation, and even differentiation at the whole organismal level. Several imaging modalities and strategies have been explored for monitoring cell grafts in vivo. We begin this review with an introduction describing the progress in stem cell technology, with a perspective toward cell replacement therapy. The importance of molecular imaging in reporting and assessing the status of cell grafts and their relation to the local microenvironment is highlighted since the current knowledge gap is one of the major obstacles in clinical translation of stem cell therapy. Based on currently available imaging techniques, we provide a brief discussion on the pros and cons of each imaging modality used for monitoring cell grafts with particular emphasis on magnetic resonance imaging (MRI) and the reporter gene approach. Finally, we conclude with a comprehensive discussion of future directions of applying molecular imaging in regenerative medicine to emphasize further the importance of correlating cell graft conditions and clinical outcomes to advance regenerative medicine.
Collapse
Affiliation(s)
- In K Cho
- Department of Human Genetics, Emory University School of MedicineAtlanta, GA, USA
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research CenterAtlanta, GA, USA
| | - Silun Wang
- Department of Radiology and Imaging Sciences, Emory University School of MedicineAtlanta, GA, USA
| | - Hui Mao
- Department of Radiology and Imaging Sciences, Emory University School of MedicineAtlanta, GA, USA
| | - Anthony WS Chan
- Department of Human Genetics, Emory University School of MedicineAtlanta, GA, USA
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research CenterAtlanta, GA, USA
| |
Collapse
|
36
|
Menezes ME, Das SK, Minn I, Emdad L, Wang XY, Sarkar D, Pomper MG, Fisher PB. Detecting Tumor Metastases: The Road to Therapy Starts Here. Adv Cancer Res 2016; 132:1-44. [PMID: 27613128 DOI: 10.1016/bs.acr.2016.07.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Metastasis is the complex process by which primary tumor cells migrate and establish secondary tumors in an adjacent or distant location in the body. Early detection of metastatic disease and effective therapeutic options for targeting these detected metastases remain impediments to effectively treating patients with advanced cancers. If metastatic lesions are identified early, patients might maximally benefit from effective early therapeutic interventions. Further, monitoring patients whose primary tumors are effectively treated for potential metastatic disease onset is also highly valuable. Finally, patients with metastatic disease can be monitored for efficacy of specific therapeutic interventions through effective metastatic detection techniques. Thus, being able to detect and visualize metastatic lesions is key and provides potential to greatly improve overall patient outcomes. In order to achieve these objectives, researchers have endeavored to mechanistically define the steps involved in the metastatic process as well as ways to effectively detect metastatic progression. We presently overview various preclinical and clinical in vitro and in vivo assays developed to more efficiently detect tumor metastases, which provides the foundation for developing more effective therapies for this invariably fatal component of the cancerous process.
Collapse
Affiliation(s)
- M E Menezes
- Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - S K Das
- Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - I Minn
- The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - L Emdad
- Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - X-Y Wang
- Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - D Sarkar
- Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - M G Pomper
- The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - P B Fisher
- Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
37
|
Wang Y, Roose BW, Palovcak EJ, Carnevale V, Dmochowski IJ. A Genetically Encoded β-Lactamase Reporter for Ultrasensitive (129) Xe NMR in Mammalian Cells. Angew Chem Int Ed Engl 2016; 55:8984-7. [PMID: 27305488 DOI: 10.1002/anie.201604055] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 05/20/2016] [Indexed: 01/27/2023]
Abstract
Molecular imaging holds considerable promise for elucidating biological processes in normal physiology as well as disease states, but requires noninvasive methods for identifying analytes at sub-micromolar concentrations. Particularly useful are genetically encoded, single-protein reporters that harness the power of molecular biology to visualize specific molecular processes, but such reporters have been conspicuously lacking for in vivo magnetic resonance imaging (MRI). Herein, we report TEM-1 β-lactamase (bla) as a single-protein reporter for hyperpolarized (HP) (129) Xe NMR, with significant saturation contrast at 0.1 μm. Xenon chemical exchange saturation transfer (CEST) interactions with the primary allosteric site in bla give rise to a unique saturation peak at 255 ppm, well removed (≈60 ppm downfield) from the (129) Xe-H2 O peak. Useful saturation contrast was also observed for bla expressed in bacterial cells and mammalian cells.
Collapse
Affiliation(s)
- Yanfei Wang
- Department of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, PA, 19104-6323, USA
| | - Benjamin W Roose
- Department of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, PA, 19104-6323, USA
| | - Eugene J Palovcak
- Institute for Computational Molecular Science, College of Science and Technology, Temple University, 1925 N. 12th Street, Philadelphia, PA, 19122, USA
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, College of Science and Technology, Temple University, 1925 N. 12th Street, Philadelphia, PA, 19122, USA
| | - Ivan J Dmochowski
- Department of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, PA, 19104-6323, USA.
| |
Collapse
|
38
|
Wang Y, Roose BW, Palovcak EJ, Carnevale V, Dmochowski IJ. A Genetically Encoded β-Lactamase Reporter for Ultrasensitive129Xe NMR in Mammalian Cells. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201604055] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Yanfei Wang
- Department of Chemistry; University of Pennsylvania; 231 South 34th Street Philadelphia PA 19104-6323 USA
| | - Benjamin W. Roose
- Department of Chemistry; University of Pennsylvania; 231 South 34th Street Philadelphia PA 19104-6323 USA
| | - Eugene J. Palovcak
- Institute for Computational Molecular Science, College of Science and Technology; Temple University; 1925 N. 12th Street Philadelphia PA 19122 USA
| | - Vincenzo Carnevale
- Institute for Computational Molecular Science, College of Science and Technology; Temple University; 1925 N. 12th Street Philadelphia PA 19122 USA
| | - Ivan J. Dmochowski
- Department of Chemistry; University of Pennsylvania; 231 South 34th Street Philadelphia PA 19104-6323 USA
| |
Collapse
|
39
|
Williford JM, Archang MM, Minn I, Ren Y, Wo M, Vandermark J, Fisher PB, Pomper MG, Mao HQ. Critical Length of PEG Grafts on lPEI/DNA Nanoparticles for Efficient in Vivo Delivery. ACS Biomater Sci Eng 2016; 2:567-578. [PMID: 27088129 PMCID: PMC4829937 DOI: 10.1021/acsbiomaterials.5b00551] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 03/02/2016] [Indexed: 12/03/2022]
Abstract
![]()
Nanoparticle-mediated
gene delivery is a promising alternative
to viral methods; however, its use in vivo, particularly following
systemic injection, has suffered from poor delivery efficiency. Although
PEGylation of nanoparticles has been successfully demonstrated as
a strategy to enhance colloidal stability, its success in improving
delivery efficiency has been limited, largely due to reduced cell
binding and uptake, leading to poor transfection efficiency. Here
we identified an optimized PEGylation scheme for DNA micellar nanoparticles
that delivers balanced colloidal stability and transfection activity.
Using linear polyethylenimine (lPEI)-g-PEG as a carrier,
we characterized the effect of graft length and density of polyethylene
glycol (PEG) on nanoparticle assembly, micelle stability, and gene
delivery efficiency. Through variation of PEG grafting degree, lPEI
with short PEG grafts (molecular weight, MW 500–700 Da) generated
micellar nanoparticles with various shapes including spherical, rodlike,
and wormlike nanoparticles. DNA micellar nanoparticles prepared with
short PEG grafts showed comparable colloidal stability in salt and
serum-containing media to those prepared with longer PEG grafts (MW
2 kDa). Corresponding to this trend, nanoparticles prepared with short
PEG grafts displayed significantly higher in vitro transfection efficiency
compared to those with longer PEG grafts. More importantly, short
PEG grafts permitted marked increase in transfection efficiency following
ligand conjugation to the PEG terminal in metastatic prostate cancer-bearing
mice. This study identifies that lPEI-g-PEG with
short PEG grafts (MW 500–700 Da) is the most effective to ensure
shape control and deliver high colloidal stability, transfection activity,
and ligand effect for DNA nanoparticles in vitro and in vivo following
intravenous administration.
Collapse
Affiliation(s)
- John-Michael Williford
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Baltimore, Maryland 21205, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Maani M Archang
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Medical Institutions , 601 N. Caroline Street, Baltimore, Maryland 21287, United States
| | - Yong Ren
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Mark Wo
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - John Vandermark
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, 1101 East Marshall Street, Richmond, Virginia 23298, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, 1220 East Broad Street, Richmond, Virginia 23298, United States; VCU Massey Cancer Center, Virginia Commonwealth University, 401 College Street, Richmond, Virginia 23298, United States
| | - Martin G Pomper
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins Medical Institutions, 601 N. Caroline Street, Baltimore, Maryland 21287, United States
| | - Hai-Quan Mao
- Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Institute for NanoBioTechnology and Department of Materials Science and Engineering, Johns Hopkins University, 3400 N. Charles Street, Baltimore, Maryland 21218, United States; Translational Tissue Engineering Center and Whitaker Biomedical Engineering Institute, Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, Maryland 21287, United States
| |
Collapse
|
40
|
Ferrauto G, Delli Castelli D, Di Gregorio E, Terreno E, Aime S. LipoCEST and cellCEST imaging agents: opportunities and challenges. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 8:602-18. [PMID: 26810631 DOI: 10.1002/wnan.1385] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/10/2015] [Accepted: 11/19/2015] [Indexed: 01/01/2023]
Abstract
From the early days of CEST agents' disclosure, it was evident that their potential for in vivo applications was strongly hampered by the intrinsic low sensitivity. Therefore, much work has been devoted to seek out suitable routes to achieve strong CEST contrast enhancement. The use of nanosized systems turned out to be a strategic choice, because a very large amount of CEST agents can be delivered at the site of interest. However, the breakthrough innovation in term of increase of sensitivity was found by designing the lipoCEST agents. The naturally inspired, liposomes vesicles, when loaded with paramagnetic lanthanide-based shift reagents, can be transformed into CEST probes. The large number of water molecules entrapped inside the inner cavity of the nanovesicles represents an enormous pool of exchanging protons for the generation of CEST contrast, whereas the presence of the shift reagent increases the separation in chemical shift of their nuclear magnetic resonance signal from that of the bulk water, thus allowing for a proper exchange regime for the activation of CEST contrast. From lipoCEST, it has been rather straightforward to evolve to cellCEST in order to exploit the cytoplasmatic water molecules as source of the CEST effect, once cells have been loaded with the proper shift reagent. The red blood cells were found to be particularly suitable for the development of the cellCEST concept. Finally, an understanding of the main determinants of the CEST effects in nanosized and cellular-sized agents has allowed the design of innovative lipoCEST/RBC aggregates for potential theranostic applications. WIREs Nanomed Nanobiotechnol 2016, 8:602-618. doi: 10.1002/wnan.1385 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Giuseppe Ferrauto
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Daniela Delli Castelli
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Enza Di Gregorio
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Enzo Terreno
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy.,IBB-CNR-UOS, University of Torino (IT), Turin, Italy
| | - Silvio Aime
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy.,IBB-CNR-UOS, University of Torino (IT), Turin, Italy
| |
Collapse
|
41
|
Daryaei I, Pagel MD. Double agents and secret agents: the emerging fields of exogenous chemical exchange saturation transfer and T 2-exchange magnetic resonance imaging contrast agents for molecular imaging. ACTA ACUST UNITED AC 2015; 5:19-32. [PMID: 27747191 PMCID: PMC5064441 DOI: 10.2147/rrnm.s81742] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Two relatively new types of exogenous magnetic resonance imaging contrast agents may provide greater impact for molecular imaging by providing greater specificity for detecting molecular imaging biomarkers. Exogenous chemical exchange saturation transfer (CEST) agents rely on the selective saturation of the magnetization of a proton on an agent, followed by chemical exchange of a proton from the agent to water. The selective detection of a biomarker-responsive CEST signal and an unresponsive CEST signal, followed by the ratiometric comparison of these signals, can improve biomarker specificity. We refer to this improvement as a "double-agent" approach to molecular imaging. Exogenous T2-exchange agents also rely on chemical exchange of protons between the agent and water, especially with an intermediate rate that lies between the slow exchange rates of CEST agents and the fast exchange rates of traditional T1 and T2 agents. Because of this intermediate exchange rate, these agents have been relatively unknown and have acted as "secret agents" in the contrast agent research field. This review exposes these secret agents and describes the merits of double agents through examples of exogenous agents that detect enzyme activity, nucleic acids and gene expression, metabolites, ions, redox state, temperature, and pH. Future directions are also provided for improving both types of contrast agents for improved molecular imaging and clinical translation. Therefore, this review provides an overview of two new types of exogenous contrast agents that are becoming useful tools within the armamentarium of molecular imaging.
Collapse
Affiliation(s)
- Iman Daryaei
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA
| | - Mark D Pagel
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, AZ, USA; Department of Biomedical engineering, University of Arizona, Tucson, AZ, USA; Department of Medical Imaging, University of Arizona, Tucson, AZ, USA; The University of Arizona Cancer Center, Tucson, AZ, USA
| |
Collapse
|
42
|
Simultaneous MR imaging for tissue engineering in a rat model of stroke. Sci Rep 2015; 5:14597. [PMID: 26419200 PMCID: PMC4588587 DOI: 10.1038/srep14597] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 09/01/2015] [Indexed: 12/31/2022] Open
Abstract
In situ tissue engineering within a stroke cavity is gradually emerging as a novel therapeutic paradigm. Considering the varied lesion topology within each subject, the placement and distribution of cells within the lesion cavity is challenging. The use of multiple cell types to reconstruct damaged tissue illustrates the complexity of the process, but also highlights the challenges to provide a non-invasive assessment. The distribution of implanted cells within the lesion cavity and crucially the contribution of neural stem cells and endothelial cells to morphogenesis could be visualized simultaneously using two paramagnetic chemical exchange saturation transfer (paraCEST) agents. The development of sophisticated imaging methods is essential to guide delivery of the building blocks for in situ tissue engineering, but will also be essential to understand the dynamics of cellular interactions leading to the formation of de novo tissue.
Collapse
|